1
|
Sumida T, Ogawa S, Higo S, Kuramoto Y, Eto R, Ikeda Y, Sun C, Li J, Liu L, Tabata T, Asano Y, Shiba M, Akazawa Y, Nakamura D, Oka T, Ohtani T, Sakata Y. Four cardiomyopathy patients with a heterozygous DSG2 p.Arg119Ter variant. Hum Genome Var 2024; 11:47. [PMID: 39706847 PMCID: PMC11661998 DOI: 10.1038/s41439-024-00304-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
DSG2, encoding desmoglein-2, is one of the causative genes of arrhythmogenic cardiomyopathy. We previously identified a homozygous DSG2 p.Arg119Ter stop-gain variant in a patient with juvenile-onset cardiomyopathy and advanced biventricular heart failure. However, the pathological significance and prevalence of the heterozygous DSG2 p.Arg119Ter variant remains uncertain. Here, we identified four unrelated patients with cardiomyopathy with heterozygous DSG2 p.Arg119Ter variants among 808 patients with nonischemic cardiomyopathy; the allele frequency was 0.0037, which is more than 50-fold greater than that reported in the general Japanese population. These patients were clinically diagnosed with arrhythmogenic right ventricular cardiomyopathy (Pt-1), dilated cardiomyopathy (DCM) after ventricular septum defect closure surgery (Pt-2), DCM (Pt-3), and end-stage hypertrophic cardiomyopathy (Pt-4). The patients also exhibited reduced left ventricular contractile function and varying clinical courses. Genetic analysis identified additional possible causative variants, DSG2 p.Arg292Cys in Pt-1 and BAG3 p.His166SerfsTer6 in Pt-3. Immunohistochemical analysis of endomyocardial biopsy samples revealed that the expression of not only desmoglein-2 but also desmoplakin was markedly reduced. Transmission electron microscopy revealed pale and fragmented desmosomes and widened gaps between intercalated discs in the myocardium. A microforce test using human cardiomyocytes differentiated from induced pluripotent stem cells (iPSC-CMs) demonstrated reduced contractility in iPSC-CMs carrying a heterozygous truncating variant in DSG2. These data suggest that the DSG2 p.Arg119Ter variant is concealed in patients with cardiomyopathy with heart failure, and desmosome impairment may be a latent exacerbating factor of contractile dysfunction and disease progression.
Collapse
Affiliation(s)
- Takuya Sumida
- Faculty of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Shou Ogawa
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Shuichiro Higo
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Ryo Eto
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoshihiko Ikeda
- Department of Pathology, National Cerebral and Cardiovascular Center, Suita, Osaka, 564-8565, Japan
| | - Congcong Sun
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Junjun Li
- Photonics Cell Evaluation Laboratory, Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Li Liu
- Photonics Cell Evaluation Laboratory, Graduate School of Engineering, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Tomoka Tabata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center, Suita, Osaka, 564-8565, Japan
| | - Mikio Shiba
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
- Cardiovascular Division, Osaka Police Hospital, Osaka, 543-0035, Japan
| | - Yasuhiro Akazawa
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Daisuke Nakamura
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Takafumi Oka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Tomohito Ohtani
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
2
|
Mo L, Sia C, Lin W, Zheng X, Peng K. Describing and Mapping the Research Trend of Scientific Publications on Arrhythmogenic Right Ventricular Cardiomyopathy Across Four Decades: A Bibliometric Analysis. Clin Cardiol 2024; 47:e70051. [PMID: 39600076 PMCID: PMC11599429 DOI: 10.1002/clc.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVES To perform a bibliometric analysis of publications of arrhythmogenic right ventricular cardiomyopathy (ARVC) from 1981 to 2023 to summarize the current publications and explore frontiers on this topic. METHODS We integrated the scientific publications on ARVC in the Web of Science (WOS) Core Collection database from January 1981 to September 2023, using the retrieval strategy of medical subject headings combined with keywords. We focused on articles and reviews that were published in English. Relevant information such as the journal and publisher, the title, authors, organizations, abstract, keywords, published date, and number of citations, were collected. Bibliometric analysis was performed and visualized by the R software and Microsoft Excel. RESULTS The results revealed a total of 4792 records related to ARVC from the WOS database, and 2992 original articles or reviews which were selected for bibliometric analysis. There were 79 countries and regions, 3724 research institutions, and 12 157 scholars who have published in this topic. The number of scientific publications of ARVC increased year-by-year, with an annual growth rate of 12.12%. We also investigated the top 10 contributing countries, organizations with affiliations, most influential researchers, highest-cited articles, and highest-frequency keywords. In addition, the most active areas of research on ARVC included that of fatal complications, molecular pathological mechanisms, diagnosis, therapy, and prognosis respectively according to the keywords trend analysis. CONCLUSIONS Our study reports the publication landscape of ARVC during the past four decades based on bibliometric analysis. This study provides a deeper understanding of the published literature on ARVC.
Collapse
Affiliation(s)
- Leitong Mo
- Department of Coronary Care UnitMaoming People's HospitalMaomingGuangdongChina
| | - Ching‐Hui Sia
- Department of CardiologyNational University Heart Centre SingaporeSingaporeSingapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Weiqin Lin
- Department of CardiologyNational University Heart Centre SingaporeSingaporeSingapore
- Department of MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Xifeng Zheng
- Department of Internal MedicineHospital of Guangdong University of TechnologyGuangzhouGuangdongChina
| | - Kaiyi Peng
- Department of Critical Care MedicineMaoming People's HospitalMaomingGuangdongChina
| |
Collapse
|
3
|
Huang S, Li J, Li Q, Wang Q, Zhou X, Chen J, Chen X, Bellou A, Zhuang J, Lei L. Cardiomyopathy: pathogenesis and therapeutic interventions. MedComm (Beijing) 2024; 5:e772. [PMID: 39465141 PMCID: PMC11502724 DOI: 10.1002/mco2.772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiomyopathy is a group of disease characterized by structural and functional damage to the myocardium. The etiologies of cardiomyopathies are diverse, spanning from genetic mutations impacting fundamental myocardial functions to systemic disorders that result in widespread cardiac damage. Many specific gene mutations cause primary cardiomyopathy. Environmental factors and metabolic disorders may also lead to the occurrence of cardiomyopathy. This review provides an in-depth analysis of the current understanding of the pathogenesis of various cardiomyopathies, highlighting the molecular and cellular mechanisms that contribute to their development and progression. The current therapeutic interventions for cardiomyopathies range from pharmacological interventions to mechanical support and heart transplantation. Gene therapy and cell therapy, propelled by ongoing advancements in overarching strategies and methodologies, has also emerged as a pivotal clinical intervention for a variety of diseases. The increasing number of causal gene of cardiomyopathies have been identified in recent studies. Therefore, gene therapy targeting causal genes holds promise in offering therapeutic advantages to individuals diagnosed with cardiomyopathies. Acting as a more precise approach to gene therapy, they are gradually emerging as a substitute for traditional gene therapy. This article reviews pathogenesis and therapeutic interventions for different cardiomyopathies.
Collapse
Affiliation(s)
- Shitong Huang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Jiaxin Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuying Li
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Qiuyu Wang
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Xianwu Zhou
- Department of Cardiovascular SurgeryZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jimei Chen
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Xuanhui Chen
- Department of Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
| | - Abdelouahab Bellou
- Department of Emergency Medicine, Institute of Sciences in Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Emergency MedicineWayne State University School of MedicineDetroitMichiganUSA
| | - Jian Zhuang
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| | - Liming Lei
- Department of Cardiac Surgical Intensive Care UnitGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityGuangzhouChina
- Department of Cardiovascular SurgeryGuangdong Provincial Key Laboratory of South China Structural Heart DiseaseGuangzhouChina
| |
Collapse
|
4
|
Newman NA, Burke MA. Dilated Cardiomyopathy: A Genetic Journey from Past to Future. Int J Mol Sci 2024; 25:11460. [PMID: 39519012 PMCID: PMC11546582 DOI: 10.3390/ijms252111460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by reduced systolic function and cardiac dilation. Cases without an identified secondary cause are classified as idiopathic dilated cardiomyopathy (IDC). Over the last 35 years, many cases of IDC have increasingly been recognized to be genetic in etiology with a core set of definitively causal genes in up to 40% of cases. While over 200 genes have been associated with DCM, the evidence supporting pathogenicity for most remains limited. Further, rapid advances in sequencing and bioinformatics have recently revealed a complex genetic spectrum ranging from monogenic to polygenic in DCM. These advances have also led to the discovery of causal and modifier genetic variants in secondary forms of DCM (e.g., alcohol-induced cardiomyopathy). Current guidelines recommend genetic counseling and screening, as well as endorsing a handful of genotype-specific therapies (e.g., device placement in LMNA cardiomyopathy). The future of genetics in DCM will likely involve polygenic risk scores, direct-to-consumer testing, and pharmacogenetics, requiring providers to have a thorough understanding of this rapidly developing field. Herein we outline three decades of genetics in DCM, summarize recent advances, and project possible future avenues for the field.
Collapse
Affiliation(s)
- Noah A. Newman
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael A. Burke
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Bufi S, Santoro R. Three-Dimensional iPSC-Based In Vitro Cardiac Models for Biomedical and Pharmaceutical Research Applications. Int J Mol Sci 2024; 25:10690. [PMID: 39409018 PMCID: PMC11477044 DOI: 10.3390/ijms251910690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular diseases are a major cause of death worldwide. Advanced in vitro models can be the key stone for a better understanding of the mechanisms at the basis of the different pathologies, supporting the development of novel therapeutic protocols. In particular, the implementation of induced pluripotent stem cell (iPSC) technology allows for the generation of a patient-specific pluripotent cell line that is able to differentiate in several organ-specific cell subsets while retaining the patient genetic background, thus putting the basis for personalized in vitro modeling toward personalized medicine. The design of iPSC-based models able to recapitulate the complexity of the cardiac environment is a critical goal. Here, we review some of the published efforts to exploit three dimensional (3D) iPSC-based methods to recapitulate the relevant cardiomyopathies, including genetically and non-genetically determined cardiomyopathies and cardiotoxicity studies. Finally, we discuss the actual method limitations and the future perspectives in the field.
Collapse
Affiliation(s)
- Simona Bufi
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, 20138 Milan, Italy
| | - Rosaria Santoro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, 20138 Milan, Italy
- Department of Electronics, Information and Biomedical Engineering, Politecnico di Milano, 20133 Milan, Italy
| |
Collapse
|
6
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
7
|
Zhang B, Wu Y, Zhou C, Xie J, Zhang Y, Yang X, Xiao J, Wang DW, Shan C, Zhou X, Xiang Y, Yang B. Hyperactivation of ATF4/TGF-β1 signaling contributes to the progressive cardiac fibrosis in Arrhythmogenic cardiomyopathy caused by DSG2 Variant. BMC Med 2024; 22:361. [PMID: 39227800 PMCID: PMC11373413 DOI: 10.1186/s12916-024-03593-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiomyopathy characterized with progressive cardiac fibrosis and heart failure. However, the exact mechanism driving the progression of cardiac fibrosis and heart failure in ACM remains elusive. This study aims to investigate the underlying mechanisms of progressive cardiac fibrosis in ACM caused by newly identified Desmoglein-2 (DSG2) variation. METHODS We identified homozygous DSG2F531C variant in a family with 8 ACM patients using whole-exome sequencing and generated Dsg2F536C knock-in mice. Neonatal and adult mouse ventricular myocytes isolated from Dsg2F536C knock-in mice were used. We performed functional, transcriptomic and mass spectrometry analyses to evaluate the mechanisms of ACM caused by DSG2F531C variant. RESULTS All eight patients with ACM were homozygous for DSG2F531C variant. Dsg2F536C/F536C mice displayed cardiac enlargement, dysfunction, and progressive cardiac fibrosis in both ventricles. Mechanistic investigations revealed that the variant DSG2-F536C protein underwent misfolding, leading to its recognition by BiP within the endoplasmic reticulum, which triggered endoplasmic reticulum stress, activated the PERK-ATF4 signaling pathway and increased ATF4 levels in cardiomyocytes. Increased ATF4 facilitated the expression of TGF-β1 in cardiomyocytes, thereby activating cardiac fibroblasts through paracrine signaling and ultimately promoting cardiac fibrosis in Dsg2F536C/F536C mice. Notably, inhibition of the PERK-ATF4 signaling attenuated progressive cardiac fibrosis and cardiac systolic dysfunction in Dsg2F536C/F536C mice. CONCLUSIONS Hyperactivation of the ATF4/TGF-β1 signaling in cardiomyocytes emerges as a novel mechanism underlying progressive cardiac fibrosis in ACM. Targeting the ATF4/TGF-β1 signaling may be a novel therapeutic target for managing ACM.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Chunjiang Zhou
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Jiaxi Xie
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Youming Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Jing Xiao
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China
| | - Dao Wu Wang
- State Key Laboratory of Reproductive Medicine, the Centre for Clinical Reproductive Medicine, Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Congjia Shan
- Model Animal Research Center, Nanjing University, Nanjing, China
| | - Xiujuan Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, P.R. China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai, 200120, P.R. China.
| |
Collapse
|
8
|
Wei M, Li Y, Liu X, Zhou K, Qiu Y, Liu L, Huang L, Liu Z. Case report: Additional variants induced sudden cardiac death among pediatric ACM with DSG2 homozygous mutant genotype: a report of three cases. Front Genet 2024; 15:1428796. [PMID: 39253717 PMCID: PMC11381389 DOI: 10.3389/fgene.2024.1428796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024] Open
Abstract
Background Mutations in genes encoding desmosomal proteins are the leading cause of arrhythmogenic cardiomyopathy (ACM). The majority of the inherited ACM cases demonstrate autosomal dominant genotype. Several cases with the homozygous DSG2 c.1592T>G (p.F531C) variant genotype demonstrate adverse clinical outcomes, but the roles of associated genetic mutations are not clear. In this report, we describe three ACM cases with the homozygous DSG2 c.1592T>G (p.F531C) variant genotype combined with additional heterozygous cardiomyopathy-related genetic mutations that cause aggravated clinical manifestations and worse clinical outcomes. Case presentation The three reported probands demonstrated similar clinical presentations such as heart failure, cardiac enlargement, and lethal arrhythmias. All of them experienced sudden cardiac death (SCD) before undergoing implantable cardioverter defibrillator (ICD) or heart transplantations. Whole-exome sequencing analysis demonstrated that the three patients inherited the homozygous DSG2 c.1592T>G (p.F531C) variant. Furthermore, probands I, II, and III also inherited additional heterozygous cardiomyopathy-associated mutations, including DSP c.7883T>C, SCN5a c.3577C>T, or MYH7 c.427C>T, respectively. These variants were confirmed as pathogenetic variants. A systematic review of all the reported ACM cases with the homozygous DSG2 variants suggested that the additional genetic mutations contributed to the early age onset of ACM and lethal cardiac events. Conclusion In conclusion, we report three rare cases of ACM with the same homozygous DSG2 variant in combination with additional heterozygous mutations in cardiomyopathy-associated genes. A systematic review of all the ACM cases with homozygous DSG2 variants demonstrated that the additional genetic variants contributed to the aggravated clinical manifestations and worse clinical symptoms of the ACM patients because of homozygous DSG2 mutations, including early disease onset and lethal cardiac events. Our data suggested that comprehensive genetic evaluation should be performed to identify any potential additional pathogenic variants that may significantly influence the clinical prognosis and outcomes of patients with ACM. The knowledge of underlying molecular mutations would be useful in designing better therapeutic strategies for ACM patients with multiple genetic mutations.
Collapse
Affiliation(s)
- Meng Wei
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaiyu Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Qiu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lili Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongqiang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Vanaja IP, Scalco A, Ronfini M, Bona AD, Olianti C, Rizzo S, Chelko SP, Corrado D, Sacconi L, Basso C, Mongillo M, Zaglia T. Cardiac sympathetic neurons are additional cells affected in genetically determined arrhythmogenic cardiomyopathy. J Physiol 2024. [PMID: 39141822 DOI: 10.1113/jp286845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/05/2024] [Indexed: 08/16/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial cardiac disease, mainly caused by mutations in desmosomal genes, which accounts for most cases of stress-related arrhythmic sudden death, in young and athletes. AC hearts display fibro-fatty lesions that generate the arrhythmic substrate and cause contractile dysfunction. A correlation between physical/emotional stresses and arrhythmias supports the involvement of sympathetic neurons (SNs) in the disease, but this has not been confirmed previously. Here, we combined molecular, in vitro and ex vivo analyses to determine the role of AC-linked DSG2 downregulation on SN biology and assess cardiac sympathetic innervation in desmoglein-2 mutant (Dsg2mut/mut) mice. Molecular assays showed that SNs express DSG2, implying that DSG2-mutation carriers would harbour the mutant protein in SNs. Confocal immunofluorescence of heart sections and 3-D reconstruction of SN network in clarified heart blocks revealed significant changes in the physiologialc SN topology, with massive hyperinnervation of the intact subepicardial layers and heterogeneous distribution of neurons in fibrotic areas. Cardiac SNs isolated from Dsg2mut/mut neonatal mice, prior to the establishment of cardiac innervation, show alterations in axonal sprouting, process development and distribution of varicosities. Consistently, virus-assisted DSG2 downregulation replicated, in PC12-derived SNs, the phenotypic alterations displayed by Dsg2mut/mut primary neurons, corroborating that AC-linked Dsg2 variants may affect SNs. Our results reveal that altered sympathetic innervation is an unrecognized feature of AC hearts, which may result from the combination of cell-autonomous and context-dependent factors implicated in myocardial remodelling. Our results favour the concept that AC is a disease of multiple cell types also hitting cardiac SNs. KEY POINTS: Arrhythmogenic cardiomyopathy is a genetically determined cardiac disease, which accounts for most cases of stress-related arrhythmic sudden death. Arrhythmogenic cardiomyopathy linked to mutations in desmoglein-2 (DSG2) is frequent and leads to a left-dominant form of the disease. Arrhythmogenic cardiomyopathy has been approached thus far as a disease of cardiomyocytes, but we here unveil that DSG2 is expressed, in addition to cardiomyocytes, by cardiac and extracardiac sympathetic neurons, although not organized into desmosomes. AC-linked DSG2 downregulation primarily affect sympathetic neurons, resulting in the significant increase in cardiac innervation density, accompanied by alterations in sympathetic neuron distribution. Our data supports the notion that AC develops with the contribution of several 'desmosomal protein-carrying' cell types and systems.
Collapse
Affiliation(s)
- Induja Perumal Vanaja
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Arianna Scalco
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Ronfini
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anna Di Bona
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Camilla Olianti
- Institute of Clinical Physiology (IFC), National Research Council, Florence, Florence, Italy
| | - Stefania Rizzo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stephen P Chelko
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- Department of Biomedical Sciences, Florida State University, College of Medicine, Tallahassee, FL, USA
| | - Domenico Corrado
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Leonardo Sacconi
- Institute of Clinical Physiology (IFC), National Research Council, Florence, Florence, Italy
- Institute for Experimental Cardiovascular Medicine, University Heart Center and Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Marco Mongillo
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Tania Zaglia
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Risato G, Brañas Casas R, Cason M, Bueno Marinas M, Pinci S, De Gaspari M, Visentin S, Rizzo S, Thiene G, Basso C, Pilichou K, Tiso N, Celeghin R. In Vivo Approaches to Understand Arrhythmogenic Cardiomyopathy: Perspectives on Animal Models. Cells 2024; 13:1264. [PMID: 39120296 PMCID: PMC11311808 DOI: 10.3390/cells13151264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a hereditary cardiac disorder characterized by the gradual replacement of cardiomyocytes with fibrous and adipose tissue, leading to ventricular wall thinning, chamber dilation, arrhythmias, and sudden cardiac death. Despite advances in treatment, disease management remains challenging. Animal models, particularly mice and zebrafish, have become invaluable tools for understanding AC's pathophysiology and testing potential therapies. Mice models, although useful for scientific research, cannot fully replicate the complexity of the human AC. However, they have provided valuable insights into gene involvement, signalling pathways, and disease progression. Zebrafish offer a promising alternative to mammalian models, despite the phylogenetic distance, due to their economic and genetic advantages. By combining animal models with in vitro studies, researchers can comprehensively understand AC, paving the way for more effective treatments and interventions for patients and improving their quality of life and prognosis.
Collapse
Affiliation(s)
- Giovanni Risato
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
- Department of Biology, University of Padua, I-35131 Padua, Italy;
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | | | - Marco Cason
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Maria Bueno Marinas
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Serena Pinci
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Monica De Gaspari
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Silvia Visentin
- Department of Women’s and Children’s Health, University of Padua, I-35128 Padua, Italy;
| | - Stefania Rizzo
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Gaetano Thiene
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Cristina Basso
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Kalliopi Pilichou
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| | - Natascia Tiso
- Department of Biology, University of Padua, I-35131 Padua, Italy;
| | - Rudy Celeghin
- Department of Cardio-Thoraco-Vascular Sciences and Public Health, University of Padua, I-35128 Padua, Italy; (G.R.); (M.C.); (M.B.M.); (S.P.); (M.D.G.); (S.R.); (G.T.); (C.B.); (K.P.); (R.C.)
| |
Collapse
|
11
|
Fabritz L, Fortmueller L, Gehmlich K, Kant S, Kemper M, Kucerova D, Syeda F, Faber C, Leube RE, Kirchhof P, Krusche CA. Endurance Training Provokes Arrhythmogenic Right Ventricular Cardiomyopathy Phenotype in Heterozygous Desmoglein-2 Mutants: Alleviation by Preload Reduction. Biomedicines 2024; 12:985. [PMID: 38790949 PMCID: PMC11117820 DOI: 10.3390/biomedicines12050985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Desmoglein-2 mutations are detected in 5-10% of patients with arrhythmogenic right ventricular cardiomyopathy (ARVC). Endurance training accelerates the development of the ARVC phenotype, leading to earlier arrhythmic events. Homozygous Dsg2 mutant mice develop a severe ARVC-like phenotype. The phenotype of heterozygous mutant (Dsg2mt/wt) or haploinsufficient (Dsg20/wt) mice is still not well understood. To assess the effects of age and endurance swim training, we studied cardiac morphology and function in sedentary one-year-old Dsg2mt/wt and Dsg20/wt mice and in young Dsg2mt/wt mice exposed to endurance swim training. Cardiac structure was only occasionally affected in aged Dsg20/wt and Dsg2mt/wt mice manifesting as small fibrotic foci and displacement of Connexin 43. Endurance swim training increased the right ventricular (RV) diameter and decreased RV function in Dsg2mt/wt mice but not in wild types. Dsg2mt/wt hearts showed increased ventricular activation times and pacing-induced ventricular arrhythmia without obvious fibrosis or inflammation. Preload-reducing therapy during training prevented RV enlargement and alleviated the electrophysiological phenotype. Taken together, endurance swim training induced features of ARVC in young adult Dsg2mt/wt mice. Prolonged ventricular activation times in the hearts of trained Dsg2mt/wt mice are therefore a potential mechanism for increased arrhythmia risk. Preload-reducing therapy prevented training-induced ARVC phenotype pointing to beneficial treatment options in human patients.
Collapse
Affiliation(s)
- Larissa Fabritz
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Lisa Fortmueller
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX1 2JD, UK
| | - Sebastian Kant
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| | - Marcel Kemper
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Dana Kucerova
- Department of Cardiology, Section of Rhythmology, University Hospital Muenster, 48149 Münster, Germany;
| | - Fahima Syeda
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
| | - Cornelius Faber
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Muenster, 48149 Münster, Germany;
| | - Rudolf E. Leube
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| | - Paulus Kirchhof
- University Center of Cardiovascular Science and Department of Cardiology, University Heart and Vascular Center, University Hospital Hamburg Eppendorf, 20246 Hamburg, Germany; (L.F.); (P.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham B15 2TT, UK; (K.G.); (M.K.); (F.S.)
| | - Claudia A. Krusche
- Institute for Molecular and Cellular Anatomy (MOCA), RWTH Aachen University, 52074 Aachen, Germany; (S.K.); (R.E.L.)
| |
Collapse
|
12
|
Blich M, Zohar Y, Cohen-Kaplan V, Minkov I, Asleh R, Horowitz-Cederboim S, Weiss K, Paperna T, Lessick J, Abadi S, Khoury A, Gepstein L, Suleiman M, Caspi O. Ser194Leu DSG2 mutation, associated with arrhythmogenic left ventricular cardiomyopathy and ventricular tachycardia. Pacing Clin Electrophysiol 2024; 47:503-510. [PMID: 38375917 DOI: 10.1111/pace.14950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Arrhythmogenic cardiomyopathy (AC) is an inherited cardiomyopathy characterized by fibro-fatty replacement of cardiomyocytes, leading to life-threatening ventricular arrhythmia and heart failure. Pathogenic variants of desmoglein2 gene (DSG2) have been reported as genetic etiologies of AC. In contrast, many reported DSG2 variants are benign or variants of uncertain significance. Correct genetic variant classification is crucial for determining the best medical therapy for the patient and family members. METHODS Pathogenicity of the DSG2 Ser194Leu variant that was identified by whole exome sequencing in a patient, who presented with ventricular tachycardia and was diagnosed with AC, was investigated by electron microscopy and immunohistochemical staining of endomyocardial biopsy sample. RESULTS Electron microscopy demonstrated a widened gap in the adhering junction and a less well-organized intercalated disk region in the mutated cardiomyocytes compared to the control. Immunohistochemical staining in the proband diagnosed with AC showed reduced expression of desmoglein 2 and connexin 43 and intercalated disc distortion. Reduced expression of DSG2 and Connexin 43 were observed in cellular cytoplasm and gap junctions. Additionally, we detected perinuclear accumulation of DSG2 and Connexin 43 in the proband sample. CONCLUSION Ser194Leu is a missense pathogenic mutation of DSG2 gene associated with arrhythmogenic left ventricular cardiomyopathy.
Collapse
Affiliation(s)
- Miry Blich
- Cardiology Division, Rambam Health Care Campus, Haifa, Israel
| | - Yaniv Zohar
- Department of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Victoria Cohen-Kaplan
- Bruce Rappaport School of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Irina Minkov
- Department of Pathology, Rambam Health Care Campus, Haifa, Israel
| | - Rabea Asleh
- Heart Institute, Hadassah Medical Center, Jerusalem, Israel
| | | | - Karin Weiss
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | - Tamar Paperna
- The Genetics Institute, Rambam Health Care Campus, Haifa, Israel
| | | | - Sobhi Abadi
- Medical Imaging Departments, Rambam Health Care Campus, Haifa, Israel
| | - Asaad Khoury
- Cardiology Division, Rambam Health Care Campus, Haifa, Israel
| | - Lior Gepstein
- Cardiology Division, Rambam Health Care Campus, Haifa, Israel
| | - Mahmud Suleiman
- Cardiology Division, Rambam Health Care Campus, Haifa, Israel
| | - Oren Caspi
- Cardiology Division, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
13
|
Galizia MS, Attili AK, Truesdell WR, Smith ED, Helms AS, Sulaiman AMA, Madamanchi C, Agarwal PP. Imaging Features of Arrhythmogenic Cardiomyopathies. Radiographics 2024; 44:e230154. [PMID: 38512728 PMCID: PMC10995833 DOI: 10.1148/rg.230154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 03/23/2024]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disease characterized by replacement of ventricular myocardium with fibrofatty tissue, predisposing the patient to ventricular arrhythmias and/or sudden cardiac death. Most cases of ACM are associated with pathogenic variants in genes that encode desmosomal proteins, an important cell-to-cell adhesion complex present in both the heart and skin tissue. Although ACM was first described as a disease predominantly of the right ventricle, it is now acknowledged that it can also primarily involve the left ventricle or both ventricles. The original right-dominant phenotype is traditionally diagnosed using the 2010 task force criteria, a multifactorial algorithm divided into major and minor criteria consisting of structural criteria based on two-dimensional echocardiographic, cardiac MRI, or right ventricular angiographic findings; tissue characterization based on endomyocardial biopsy results; repolarization and depolarization abnormalities based on electrocardiographic findings; arrhythmic features; and family history. Shortfalls in the task force criteria due to the modern understanding of the disease have led to development of the Padua criteria, which include updated criteria for diagnosis of the right-dominant phenotype and new criteria for diagnosis of the left-predominant and biventricular phenotypes. In addition to incorporating cardiac MRI findings of ventricular dilatation, systolic dysfunction, and regional wall motion abnormalities, the new Padua criteria emphasize late gadolinium enhancement at cardiac MRI as a key feature in diagnosis and imaging-based tissue characterization. Conditions to consider in the differential diagnosis of the right-dominant phenotype include various other causes of right ventricular dilatation such as left-to-right shunts and variants of normal right ventricular anatomy that can be misinterpreted as abnormalities. The left-dominant phenotype can mimic myocarditis at imaging and clinical examination. Additional considerations for the differential diagnosis of ACM, particularly for the left-dominant phenotype, include sarcoidosis and dilated cardiomyopathy. ©RSNA, 2024 Test Your Knowledge questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Mauricio S Galizia
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Anil K Attili
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - William R Truesdell
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Eric D Smith
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Adam S Helms
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Abdulbaset M A Sulaiman
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Chaitanya Madamanchi
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| | - Prachi P Agarwal
- From the Department of Radiology (M.S.G., A.K.A., W.R.T., P.P.A.) and Division of Cardiovascular Medicine, Department of Internal Medicine (E.D.S., A.S.H., A.M.A.S., C.M.), Michigan Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI 48109
| |
Collapse
|
14
|
Celeghin R, Risato G, Beffagna G, Cason M, Bueno Marinas M, Della Barbera M, Facchinello N, Giuliodori A, Brañas Casas R, Caichiolo M, Vettori A, Grisan E, Rizzo S, Dalla Valle L, Argenton F, Thiene G, Tiso N, Pilichou K, Basso C. A novel DSP zebrafish model reveals training- and drug-induced modulation of arrhythmogenic cardiomyopathy phenotypes. Cell Death Discov 2023; 9:441. [PMID: 38057295 DOI: 10.1038/s41420-023-01741-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 10/30/2023] [Accepted: 11/23/2023] [Indexed: 12/08/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an inherited disorder characterized by progressive loss of the ventricular myocardium causing life-threatening ventricular arrhythmias, syncope and sudden cardiac death in young and athletes. About 40% of AC cases carry one or more mutations in genes encoding for desmosomal proteins, including Desmoplakin (Dsp). We present here the first stable Dsp knock-out (KO) zebrafish line able to model cardiac alterations and cell signalling dysregulation, characteristic of the AC disease, on which environmental factors and candidate drugs can be tested. Our stable Dsp knock-out (KO) zebrafish line was characterized by cardiac alterations, oedema and bradycardia at larval stages. Histological analysis of mutated adult hearts showed reduced contractile structures and abnormal shape of the ventricle, with thinning of the myocardial layer, vessels dilation and presence of adipocytes within the myocardium. Moreover, TEM analysis revealed "pale", disorganized and delocalized desmosomes. Intensive physical training protocol caused a global worsening of the cardiac phenotype, accelerating the progression of the disease. Of note, we detected a decrease of Wnt/β-catenin signalling, recently associated with AC pathogenesis, as well as Hippo/YAP-TAZ and TGF-β pathway dysregulation. Pharmacological treatment of mutated larvae with SB216763, a Wnt/β-catenin agonist, rescued pathway expression and cardiac abnormalities, stabilizing the heart rhythm. Overall, our Dsp KO zebrafish line recapitulates many AC features observed in human patients, pointing at zebrafish as a suitable system for in vivo analysis of environmental modulators, such as the physical exercise, and the screening of pathway-targeted drugs, especially related to the Wnt/β-catenin signalling cascade.
Collapse
Affiliation(s)
- Rudy Celeghin
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Giovanni Risato
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Giorgia Beffagna
- Department of Biology, University of Padova, Padova, 35131, Italy.
| | - Marco Cason
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Maria Bueno Marinas
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Mila Della Barbera
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Nicola Facchinello
- Neuroscience Institute, Italian National Research Council (CNR), Padova, 35131, Italy
| | - Alice Giuliodori
- Department of Biology, University of Padova, Padova, 35131, Italy
| | | | - Micol Caichiolo
- Department of Biology, University of Padova, Padova, 35131, Italy
| | - Andrea Vettori
- Department of Biotechnology, University of Verona, Verona, 37134, Italy
| | - Enrico Grisan
- School of Engineering, London South Bank University, London, SE1 0AA, UK
| | - Stefania Rizzo
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | | | | | - Gaetano Thiene
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Natascia Tiso
- Department of Biology, University of Padova, Padova, 35131, Italy.
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy
| | - Cristina Basso
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, 35128, Italy.
| |
Collapse
|
15
|
Toro V, Jutras-Beaudoin N, Boucherat O, Bonnet S, Provencher S, Potus F. Right Ventricle and Epigenetics: A Systematic Review. Cells 2023; 12:2693. [PMID: 38067121 PMCID: PMC10705252 DOI: 10.3390/cells12232693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
There is an increasing recognition of the crucial role of the right ventricle (RV) in determining the functional status and prognosis in multiple conditions. In the past decade, the epigenetic regulation (DNA methylation, histone modification, and non-coding RNAs) of gene expression has been raised as a critical determinant of RV development, RV physiological function, and RV pathological dysfunction. We thus aimed to perform an up-to-date review of the literature, gathering knowledge on the epigenetic modifications associated with RV function/dysfunction. Therefore, we conducted a systematic review of studies assessing the contribution of epigenetic modifications to RV development and/or the progression of RV dysfunction regardless of the causal pathology. English literature published on PubMed, between the inception of the study and 1 January 2023, was evaluated. Two authors independently evaluated whether studies met eligibility criteria before study results were extracted. Amongst the 817 studies screened, 109 studies were included in this review, including 69 that used human samples (e.g., RV myocardium, blood). While 37 proposed an epigenetic-based therapeutic intervention to improve RV function, none involved a clinical trial and 70 are descriptive. Surprisingly, we observed a substantial discrepancy between studies investigating the expression (up or down) and/or the contribution of the same epigenetic modifications on RV function or development. This exhaustive review of the literature summarizes the relevant epigenetic studies focusing on RV in human or preclinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | - François Potus
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec (CRIUCPQ), Québec, QC G1V 4G5, Canada; (V.T.); (N.J.-B.); (O.B.); (S.B.); (S.P.)
| |
Collapse
|
16
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
17
|
Zhang B, Wu Y, Yang X, Xiang Y, Yang B. Molecular insight into arrhythmogenic cardiomyopathy caused by DSG2 mutations. Biomed Pharmacother 2023; 167:115448. [PMID: 37696084 DOI: 10.1016/j.biopha.2023.115448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/13/2023] Open
Abstract
Mutant desmoglein 2 (DSG2) is the second most common pathogenic gene in arrhythmogenic cardiomyopathy (ACM), accounting for approximately 10% of ACM cases. In addition to common clinical and pathological features, ACM caused by mutant DSG2 has specific characteristics, manifesting as left ventricle involvement and a high risk of heart failure. Pathological studies have shown extensive cardiomyocyte necrosis, infiltration of immune cells, and fibrofatty replacement in both ventricles, as well as abnormal desmosome structures in the hearts of humans and mice with mutant DSG2-related ACM. Although desmosome dysfunction is a common pathway in the pathogenesis of mutant DSG2-related ACM, the mechanisms underlying this dysfunction vary among mutations. Desmosome dysfunction induces cardiomyocyte injury, plakoglobin dislocation, and gap junction dysfunction, all of which contribute to the initiation and progression of ACM. Additionally, dysregulated inflammation, overactivation of transforming growth factor-beta-1 signaling and endoplasmic reticulum stress, and cardiac metabolic dysfunction contribute to the pathogenesis of ACM caused by mutant DSG2. These features demonstrate that patients with mutant DSG2-related ACM should be managed individually and precisely based on the genotype and phenotype. Further studies are needed to investigate the underlying mechanisms and to identify novel therapies to reverse or attenuate the progression of ACM caused by mutant DSG2.
Collapse
Affiliation(s)
- Baowei Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yizhang Wu
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Xingbo Yang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China
| | - Yaozu Xiang
- Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Pudong, Shanghai 200120, PR China.
| |
Collapse
|
18
|
Tadros HJ, Miyake CY, Kearney DL, Kim JJ, Denfield SW. The Many Faces of Arrhythmogenic Cardiomyopathy: An Overview. Appl Clin Genet 2023; 16:181-203. [PMID: 37933265 PMCID: PMC10625769 DOI: 10.2147/tacg.s383446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a disease that involves electromechanical uncoupling of cardiomyocytes. This leads to characteristic histologic changes that ultimately lead to the arrhythmogenic clinical features of the disease. Initially thought to affect the right ventricle predominantly, more recent data show that it can affect both the ventricles or the left ventricle alone. Throughout the recent era, diagnostic modalities and criteria for AC have continued to evolve and our understanding of its clinical features in different age groups as well as the genotype to the phenotype correlations have improved. In this review, we set out to detail the epidemiology, etiologies, presentations, evaluation, and management of AC across the age continuum.
Collapse
Affiliation(s)
- Hanna J Tadros
- Department of Pediatrics, Section of Pediatric Cardiology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Christina Y Miyake
- Department of Pediatrics, Section of Pediatric Cardiology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Debra L Kearney
- Department of Pathology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey J Kim
- Department of Pediatrics, Section of Pediatric Cardiology, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Susan W Denfield
- Department of Pediatrics, Division of Pediatric Cardiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Bernini Di Michele A, Onofri V, Pesaresi M, Turchi C. The Role of miRNA Expression Profile in Sudden Cardiac Death Cases. Genes (Basel) 2023; 14:1954. [PMID: 37895303 PMCID: PMC10606010 DOI: 10.3390/genes14101954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Sudden cardiac death (SCD) is one of the leading causes of death in the world and for this reason it has attracted the attention of numerous researchers in the field of legal medicine. It is not easy to determine the cause in a SCD case and the available methods used for diagnosis cannot always give an exhaustive answer. In addition, the molecular analysis of genes does not lead to a clear conclusion, but it could be interesting to focus attention on the expression level of miRNAs, a class of non-coding RNA of about 22 nucleotides. The role of miRNAs is to regulate the gene expression through complementary binding to 3'-untraslated regions of miRNAs, leading to the inhibition of translation or to mRNA degradation. In recent years, several studies were performed with the aim of exploring the use of these molecules as biomarkers for SCD cases, and to also distinguish the causes that lead to cardiac death. In this review, we summarize experiments, evidence, and results of different studies on the implication of miRNAs in SCD cases. We discuss the different biological starting materials with their respective advantages and disadvantages, studying miRNA expression on tissue (fresh-frozen tissue and FFPE tissue), circulating cell-free miRNAs in blood of patients affected by cardiac disease at high risk of SCD, and exosomal miRNAs analyzed from serum of people who died from SCD.
Collapse
Affiliation(s)
- Alessia Bernini Di Michele
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Valerio Onofri
- Legal Medicine Unit, AOU Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy;
| | - Mauro Pesaresi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Chiara Turchi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| |
Collapse
|
20
|
Al-Ghamdi BS, Alhadeq F, Alqahtani A, Alruwaili N, Rababh M, Alghamdi S, Almanea W, Alhassnan Z. Clinical and Genetic Characteristics of Arrhythmogenic Right Ventricular Cardiomyopathy Patients: A Single-Center Experience. Cardiol Res 2023; 14:379-386. [PMID: 37936624 PMCID: PMC10627368 DOI: 10.14740/cr1531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/22/2023] [Indexed: 11/09/2023] Open
Abstract
Background Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited progressive cardiomyopathy. We aimed to define the long-term clinical outcome and genetic characteristics of patients and family members with positive genetic tests for ARVC in a single tertiary care cardiac center in Saudi Arabia. Methods We enrolled 46 subjects in the study, including 23 index-patients (probands) with ARVC based on the revised 2010 ARVC Task Force Criteria (TFC) and 23 family members who underwent a genetic test for the ARVC between 2016 and 2020. Results Of the probands, 17 (73.9%) were males with a mean age at presentation of 24.95 ± 13.9 years (7 to 55 years). Predominant symptoms were palpitations in 14 patients (60.9%), and syncope in 10 patients (43.47%). Sustained ventricular tachycardia (VT) was documented in 12 patients (52.2%). The mean left ventricular ejection fraction (LVEF) by echocardiogram was 52.81±6.311% (30-55%), and the mean right ventricular ejection fraction (RVEF) by cardiac MRI was 41.3±11.37% (23-64%). Implantable cardioverter-defibrillator (ICD) implantation was performed in 17 patients (73.9%), and over a mean follow-up of 13.65 ± 6.83 years, appropriate ICD therapy was noted in 12 patients (52.2%). Genetic variants were identified in 33 subjects (71.7%), 16 patients and 17 family members, with the most common variant of plakophilin 2 (PKP2) in 27 subjects (81.8%). Conclusions ARVC occurs during early adulthood in Saudi patients. It is associated with a significant arrhythmia burden in these patients. The PKP2 gene is the most common gene defect in Saudi patients, consistent with what is observed in other nations. We reported in this study two novel variants in PKP2 and desmocollin 2 (DSC2) genes. Genetic counseling is needed to include all first-degree family members for early diagnosis and management of the disease in our country.
Collapse
Affiliation(s)
- Bandar Saeed Al-Ghamdi
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Faten Alhadeq
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| | - Aisha Alqahtani
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| | - Nadiah Alruwaili
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
| | | | | | - Waleed Almanea
- Heart Centre Department, King Faisal Specialist Hospital & Research Center (KFSH&RC), Riyadh, Saudi Arabia
- Pediatric Cardiology, Security Forces Hospital, Riyadh, Saudi Arabia
| | - Zuhair Alhassnan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Cardiovascular Genetics Program, Center for Genomic Medicine, King Faisal Specialist Hospital & Research Centre (KFSH&RC), Riyadh, Saudi Arabia
| |
Collapse
|
21
|
Uchechukwu CF, Anyaduba UL, Udekwu CC, Orababa OQ, Kade AE. Desmoglein-2 and COVID-19 complications: insights into its role as a biomarker, pathogenesis and clinical implications. J Gen Virol 2023; 104. [PMID: 37815458 DOI: 10.1099/jgv.0.001902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023] Open
Abstract
Desmoglein-2 (DSG2) has emerged as a potential biomarker for coronavirus disease 2019 (COVID-19) complications, particularly cardiac and cardiovascular involvement. The expression of DSG2 in lung tissues has been detected at elevated levels, and circulating DSG2 levels correlate with COVID-19 severity. DSG2 may contribute to myocardial injury, cardiac dysfunction and vascular endothelial dysfunction in COVID-19. Monitoring DSG2 levels could aid in risk stratification, early detection and prognostication of COVID-19 complications. However, further research is required to validate DSG2 as a biomarker. Such research will aim to elucidate its precise role in pathogenesis, establishing standardized assays for its measurement and possibly identifying therapeutic targets.
Collapse
Affiliation(s)
- Chidiebere F Uchechukwu
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Warwick Medical School, University of Warwick, Coventry, UK
- School of Life Sciences, University of Warwick, Coventry, UK
- Michael Okpara University of Agriculture, Umudike, Nigeria
| | | | | | | | | |
Collapse
|
22
|
Chua CJ, Morrissette-McAlmon J, Tung L, Boheler KR. Understanding Arrhythmogenic Cardiomyopathy: Advances through the Use of Human Pluripotent Stem Cell Models. Genes (Basel) 2023; 14:1864. [PMID: 37895213 PMCID: PMC10606441 DOI: 10.3390/genes14101864] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiomyopathies (CMPs) represent a significant healthcare burden and are a major cause of heart failure leading to premature death. Several CMPs are now recognized to have a strong genetic basis, including arrhythmogenic cardiomyopathy (ACM), which predisposes patients to arrhythmic episodes. Variants in one of the five genes (PKP2, JUP, DSC2, DSG2, and DSP) encoding proteins of the desmosome are known to cause a subset of ACM, which we classify as desmosome-related ACM (dACM). Phenotypically, this disease may lead to sudden cardiac death in young athletes and, during late stages, is often accompanied by myocardial fibrofatty infiltrates. While the pathogenicity of the desmosome genes has been well established through animal studies and limited supplies of primary human cells, these systems have drawbacks that limit their utility and relevance to understanding human disease. Human induced pluripotent stem cells (hiPSCs) have emerged as a powerful tool for modeling ACM in vitro that can overcome these challenges, as they represent a reproducible and scalable source of cardiomyocytes (CMs) that recapitulate patient phenotypes. In this review, we provide an overview of dACM, summarize findings in other model systems linking desmosome proteins with this disease, and provide an up-to-date summary of the work that has been conducted in hiPSC-cardiomyocyte (hiPSC-CM) models of dACM. In the context of the hiPSC-CM model system, we highlight novel findings that have contributed to our understanding of disease and enumerate the limitations, prospects, and directions for research to consider towards future progress.
Collapse
Affiliation(s)
- Christianne J. Chua
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Justin Morrissette-McAlmon
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
| | - Kenneth R. Boheler
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.J.C.); (J.M.-M.); (L.T.)
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
23
|
Kurzlechner LM, Kishnani S, Chowdhury S, Atkins SL, Moya-Mendez ME, Parker LE, Rosamilia MB, Tadros HJ, Pace LA, Patel V, Chahal CAA, Landstrom AP. DiscoVari: A Web-Based Precision Medicine Tool for Predicting Variant Pathogenicity in Cardiomyopathy- and Channelopathy-Associated Genes. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:317-327. [PMID: 37409478 PMCID: PMC10527712 DOI: 10.1161/circgen.122.003911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 05/30/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND With genetic testing advancements, the burden of incidentally identified cardiac disease-associated gene variants is rising. These variants may carry a risk of sudden cardiac death, highlighting the need for accurate diagnostic interpretation. We sought to identify pathogenic hotspots in sudden cardiac death-associated genes using amino acid-level signal-to-noise (S:N) analysis and develop a web-based precision medicine tool, DiscoVari, to improve variant evaluation. METHODS The minor allele frequency of putatively pathogenic variants was derived from cohort-based cardiomyopathy and channelopathy studies in the literature. We normalized disease-associated minor allele frequencies to rare variants in an ostensibly healthy population (Genome Aggregation Database) to calculate amino acid-level S:N. Amino acids with S:N above the gene-specific threshold were defined as hotspots. DiscoVari was built using JavaScript ES6 and using open-source JavaScript library ReactJS, web development framework Next.js, and JavaScript runtime NodeJS. We validated the ability of DiscoVari to identify pathogenic variants using variants from ClinVar and individuals clinically evaluated at the Duke University Hospitals with cardiac genetic testing. RESULTS We developed DiscoVari as an internet-based tool for S:N-based variant hotspots. Upon validation, a higher proportion of ClinVar likely pathogenic/pathogenic variants localized to DiscoVari hotspots (43.1%) than likely benign/benign variants (17.8%; P<0.0001). Further, 75.3% of ClinVar variants reclassified to likely pathogenic/pathogenic were in hotspots, compared with 41.3% of those reclassified as variants of uncertain significance (P<0.0001) and 23.4% of those reclassified as likely benign/benign (P<0.0001). Of the clinical cohort variants, 73.1% of likely pathogenic/pathogenic were in hotspots, compared with 0.0% of likely benign/benign (P<0.01). CONCLUSIONS DiscoVari reliably identifies disease-susceptible amino acid residues to evaluate variants by searching amino acid-specific S:N ratios.
Collapse
Affiliation(s)
| | - Sujata Kishnani
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
| | - Shawon Chowdhury
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
| | - Sage L. Atkins
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
| | | | - Lauren E. Parker
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
| | | | - Hanna J. Tadros
- Dept of Pediatrics, Section of Pediatric Cardiology, Baylor College of Medicine, Houston, TX
| | - Leslie A. Pace
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
| | - Viraj Patel
- North West Thames Regional Genetics Service, St Mark’s Hospital, London, United Kingdom
| | - C. Anwar A. Chahal
- Center for Inherited Cardiovascular Diseases, WellSpan Health, Lancaster, PA
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
- Cardiac Electrophysiology, Cardiovascular Division, Hospital of the Univ of Pennsylvania, Philadelphia, PA
- Dept of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Andrew P. Landstrom
- Dept of Pediatrics, Division of Pediatric Cardiology, Durham, NC
- Dept of Cell Biology, Duke Univ School of Medicine, Durham, NC
| |
Collapse
|
24
|
Ward KE, Steadman L, Karim AR, Reynolds GM, Pugh M, Chua W, Faustini SE, Veenith T, Thwaites RS, Openshaw PJM, Drayson MT, Shields AM, Cunningham AF, Wraith DC, Richter AG. SARS-CoV-2 infection is associated with anti-desmoglein 2 autoantibody detection. Clin Exp Immunol 2023; 213:243-251. [PMID: 37095599 PMCID: PMC10651225 DOI: 10.1093/cei/uxad046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 04/26/2023] Open
Abstract
Post-acute cardiac sequelae, following SARS-CoV-2 infection, are well recognized as complications of COVID-19. We have previously shown the persistence of autoantibodies against antigens in skin, muscle, and heart in individuals following severe COVID-19; the most common staining on skin tissue displayed an inter-cellular cement pattern consistent with antibodies against desmosomal proteins. Desmosomes play a critical role in maintaining the structural integrity of tissues. For this reason, we analyzed desmosomal protein levels and the presence of anti-desmoglein (DSG) 1, 2, and 3 antibodies in acute and convalescent sera from patients with COVID-19 of differing clinical severity. We find increased levels of DSG2 protein in sera from acute COVID-19 patients. Furthermore, we find that DSG2 autoantibody levels are increased significantly in convalescent sera following severe COVID-19 but not in hospitalized patients recovering from influenza infection or healthy controls. Levels of autoantibody in sera from patients with severe COVID-19 were comparable to levels in patients with non-COVID-19-associated cardiac disease, potentially identifying DSG2 autoantibodies as a novel biomarker for cardiac damage. To determine if there was any association between severe COVID-19 and DSG2, we stained post-mortem cardiac tissue from patients who died from COVID-19 infection. This confirmed DSG2 protein within the intercalated discs and disruption of the intercalated disc between cardiomyocytes in patients who died from COVID-19. Our results reveal the potential for DSG2 protein and autoimmunity to DSG2 to contribute to unexpected pathologies associated with COVID-19 infection.
Collapse
Affiliation(s)
- Kerensa E Ward
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Lora Steadman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Abid R Karim
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Gary M Reynolds
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Matthew Pugh
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Winnie Chua
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, West Midlands, UK
| | - Sian E Faustini
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Tonny Veenith
- Department of Critical Care, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Mark T Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Adrian M Shields
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Adam F Cunningham
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Alex G Richter
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Clinical Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
25
|
Thiene G, Basso C, Pilichou K, Bueno Marinas M. Desmosomal Arrhythmogenic Cardiomyopathy: The Story Telling of a Genetically Determined Heart Muscle Disease. Biomedicines 2023; 11:2018. [PMID: 37509658 PMCID: PMC10377062 DOI: 10.3390/biomedicines11072018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The history of arrhythmogenic cardiomyopathy (AC) as a genetically determined desmosomal disease started since the original discovery by Lancisi in a four-generation family, published in 1728. Contemporary history at the University of Padua started with Dalla Volta, who haemodynamically investigated patients with "auricularization" of the right ventricle, and with Nava, who confirmed familiarity. The contemporary knowledge advances consisted of (a) AC as a heart muscle disease with peculiar electrical instability of the right ventricle; (b) the finding of pathological substrates, in keeping with a myocardial dystrophy; (c) the inclusion of AC in the cardiomyopathies classification; (d) AC as the main cause of sudden death in athletes; (e) the discovery of the culprit genes coding proteins of the intercalated disc (desmosome); (f) progression in clinical diagnosis with specific ECG abnormalities, angiocardiography, endomyocardial biopsy, 2D echocardiography, electron anatomic mapping and cardiac magnetic resonance; (g) the discovery of left ventricular AC; (h) prevention of SCD with the invention and application of the lifesaving implantable cardioverter defibrillator and external defibrillator scattered in public places and playgrounds as well as the ineligibility for competitive sport activity for AC patients; (i) genetic screening of the proband family to unmask asymptomatic carriers. Nondesmosomal ACs, with a phenotype overlapping desmosomal AC, are also treated, including genetics: Transmembrane protein 43, SCN5A, Desmin, Phospholamban, Lamin A/C, Filamin C, Cadherin 2, Tight junction protein 1.
Collapse
Affiliation(s)
- Gaetano Thiene
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| | - Maria Bueno Marinas
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Medical School, University of Padua, 35121 Padova, Italy
| |
Collapse
|
26
|
Htet M, Lei S, Bajpayi S, Zoitou A, Chamakioti M, Tampakakis E. The role of noncoding genetic variants in cardiomyopathy. Front Cardiovasc Med 2023; 10:1116925. [PMID: 37283586 PMCID: PMC10239966 DOI: 10.3389/fcvm.2023.1116925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
Cardiomyopathies remain one of the leading causes of morbidity and mortality worldwide. Environmental risk factors and genetic predisposition account for most cardiomyopathy cases. As with all complex diseases, there are significant challenges in the interpretation of the molecular mechanisms underlying cardiomyopathy-associated genetic variants. Given the technical improvements and reduced costs of DNA sequence technologies, an increasing number of patients are now undergoing genetic testing, resulting in a continuously expanding list of novel mutations. However, many patients carry noncoding genetic variants, and although emerging evidence supports their contribution to cardiac disease, their role in cardiomyopathies remains largely understudied. In this review, we summarize published studies reporting on the association of different types of noncoding variants with various types of cardiomyopathies. We focus on variants within transcriptional enhancers, promoters, intronic sites, and untranslated regions that are likely associated with cardiac disease. Given the broad nature of this topic, we provide an overview of studies that are relatively recent and have sufficient evidence to support a significant degree of causality. We believe that more research with additional validation of noncoding genetic variants will provide further mechanistic insights on the development of cardiac disease, and noncoding variants will be increasingly incorporated in future genetic screening tests.
Collapse
Affiliation(s)
- Myo Htet
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Shunyao Lei
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Sheetal Bajpayi
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
| | - Asimina Zoitou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | | | - Emmanouil Tampakakis
- Department of Medicine, Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
27
|
Using Zebrafish Animal Model to Study the Genetic Underpinning and Mechanism of Arrhythmogenic Cardiomyopathy. Int J Mol Sci 2023; 24:ijms24044106. [PMID: 36835518 PMCID: PMC9966228 DOI: 10.3390/ijms24044106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is largely an autosomal dominant genetic disorder manifesting fibrofatty infiltration and ventricular arrhythmia with predominantly right ventricular involvement. ACM is one of the major conditions associated with an increased risk of sudden cardiac death, most notably in young individuals and athletes. ACM has strong genetic determinants, and genetic variants in more than 25 genes have been identified to be associated with ACM, accounting for approximately 60% of ACM cases. Genetic studies of ACM in vertebrate animal models such as zebrafish (Danio rerio), which are highly amenable to large-scale genetic and drug screenings, offer unique opportunities to identify and functionally assess new genetic variants associated with ACM and to dissect the underlying molecular and cellular mechanisms at the whole-organism level. Here, we summarize key genes implicated in ACM. We discuss the use of zebrafish models, categorized according to gene manipulation approaches, such as gene knockdown, gene knock-out, transgenic overexpression, and CRISPR/Cas9-mediated knock-in, to study the genetic underpinning and mechanism of ACM. Information gained from genetic and pharmacogenomic studies in such animal models can not only increase our understanding of the pathophysiology of disease progression, but also guide disease diagnosis, prognosis, and the development of innovative therapeutic strategies.
Collapse
|
28
|
Reactivation of PPAR α alleviates myocardial lipid accumulation and cardiac dysfunction by improving fatty acid β-oxidation in Dsg2-deficient arrhythmogenic cardiomyopathy. Acta Pharm Sin B 2023; 13:192-203. [PMID: 36815030 PMCID: PMC9939300 DOI: 10.1016/j.apsb.2022.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/27/2022] [Accepted: 04/02/2022] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM), a fatal heart disease characterized by fibroadipocytic replacement of cardiac myocytes, accounts for 20% of sudden cardiac death and lacks effective treatment. It is often caused by mutations in desmosome proteins, with Desmoglein-2 (DSG2) mutations as a common etiology. However, the mechanism underlying the accumulation of fibrofatty in ACM remains unknown, which impedes the development of curative treatment. Here we investigated the fat accumulation and the underlying mechanism in a mouse model of ACM induced by cardiac-specific knockout of Dsg2 (CS-Dsg2 -/-). Heart failure and cardiac lipid accumulation were observed in CS-Dsg2 -/- mice. We demonstrated that these phenotypes were caused by decline of fatty acid (FA) β-oxidation resulted from impaired mammalian target of rapamycin (mTOR) signaling. Rapamycin worsened while overexpression of mTOR and 4EBP1 rescued the FA β-oxidation pathway in CS-Dsg2 -/- mice. Reactivation of PPARα by fenofibrate or AAV9-Pparα significantly alleviated the lipid accumulation and restored cardiac function. Our results suggest that impaired mTOR-4EBP1-PPARα-dependent FA β-oxidation contributes to myocardial lipid accumulation in ACM and PPARα may be a potential target for curative treatment of ACM.
Collapse
|
29
|
Qiu Z, Zhao Y, Tao T, Guo W, Liu R, Huang J, Xu G. Activation of PPARα Ameliorates Cardiac Fibrosis in Dsg2-Deficient Arrhythmogenic Cardiomyopathy. Cells 2022; 11:3184. [PMID: 36291052 PMCID: PMC9601208 DOI: 10.3390/cells11203184] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Arrhythmogenic cardiomyopathy (ACM) is a genetic heart muscle disease characterized by progressive fibro-fatty replacement of cardiac myocytes. Up to now, the existing therapeutic modalities for ACM are mostly palliative. About 50% of ACM is caused by mutations in genes encoding desmosomal proteins including Desmoglein-2 (Dsg2). In the current study, the cardiac fibrosis of ACM and its underlying mechanism were investigated by using a cardiac-specific knockout of Dsg2 mouse model. METHODS Cardiac-specific Dsg2 knockout (CS-Dsg2-/-) mice and wild-type (WT) mice were respectively used as the animal model of ACM and controls. The myocardial collagen volume fraction was determined by histological analysis. The expression levels of fibrotic markers such as α-SMA and Collagen I as well as signal transducers such as STAT3, SMAD3, and PPARα were measured by Western blot and quantitative real-time PCR. RESULTS Increased cardiac fibrosis was observed in CS-Dsg2-/- mice according to Masson staining. PPARα deficiency and hyperactivation of STAT3 and SMAD3 were observed in the myocardium of CS-Dsg2-/- mice. The biomarkers of fibrosis such as α-SMA and Collagen I were upregulated after gene silencing of Dsg2 in HL-1 cells. Furthermore, STAT3 gene silencing by Stat3 siRNA inhibited the expression of fibrotic markers. The activation of PPARα by fenofibrate or AAV9-Pparα improved the cardiac fibrosis and decreased the phosphorylation of STAT3, SMAD3, and AKT in CS-Dsg2-/- mice. CONCLUSIONS Activation of PPARα alleviates the cardiac fibrosis in ACM.
Collapse
Affiliation(s)
- Zirui Qiu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Yawen Zhao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Tian Tao
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Wenying Guo
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Ruonan Liu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Jingmin Huang
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
| | - Geyang Xu
- Department of Physiology, School of Medicine, Jinan University, 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, China
- Center for Clinical Epidemiology and Methodology (CCEM), Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| |
Collapse
|
30
|
Zhang N, Wang S, Wong CC. Proteomics research of SARS-CoV-2 and COVID-19 disease. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:427-445. [PMID: 37724330 PMCID: PMC10388787 DOI: 10.1515/mr-2022-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/06/2022] [Indexed: 09/20/2023]
Abstract
Currently, coronavirus disease 2019 (COVID-19) is still spreading in a global scale, exerting a massive health and socioeconomic crisis. Deep insights into the molecular functions of the viral proteins and the pathogenesis of this infectious disease are urgently needed. In this review, we comprehensively describe the proteome of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and summarize their protein interaction map with host cells. In the protein interaction network between the virus and the host, a total of 787 host prey proteins that appeared in at least two studies or were verified by co-immunoprecipitation experiments. Together with 29 viral proteins, a network of 1762 proximal interactions were observed. We also review the proteomics results of COVID-19 patients and proved that SARS-CoV-2 hijacked the host's translation system, post-translation modification system, and energy supply system via viral proteins, resulting in various immune disorders, multiple cardiomyopathies, and cholesterol metabolism diseases.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Siyuan Wang
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
| | - Catherine C.L. Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, P. R. China
- Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, P. R. China
| |
Collapse
|
31
|
Hung PF, Chung FP, Hung CL, Lin YJ, Kuo TT, Liao JN, Chen YY, Pan CH, Shaw KP, Chen SA. Decreased Expression of Plakophilin-2 and αT-Catenin in Arrhythmogenic Right Ventricular Cardiomyopathy: Potential Markers for Diagnosis. Int J Mol Sci 2022; 23:ijms23105529. [PMID: 35628349 PMCID: PMC9141850 DOI: 10.3390/ijms23105529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/01/2023] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a hereditary disease of the heart muscle. Clinical challenges remain, however, in identifying patients with ARVC in the early or concealed stages with subtle clinical manifestations. Therefore, we wanted to identify potential targets by immunohistochemical (IHC) analysis in comparison with controls. Pathogenic mutations were identified in 11 of 37 autopsied patients with ARVC. As observed from IHC analysis of the RV, expression of αT-catenin and plakophilin-2 is significantly decreased in autopsied patients with ARVC as compared to controls, and the decreased expression is consistent in patients with and without pathogenic mutations. Furthermore, ARVC specimens demonstrated a reduced localization of αT-catenin, desmocollin-2, desmoglein-2, desmoplakin, and plakophilin-2 on intercalated discs. These findings have been validated by comparing RV specimens obtained via endomyocardial biopsy between patients with ARVC and those without. The pathogenic mutation was present in 3 of 5 clinical patients with ARVC. In HL-1 myocytes, siRNA was used to knockdown CTNNA3, and western blotting analysis demonstrated that the decline in αT-catenin expression was accompanied by a significant decline in the expression of plakophilin-2. The aforementioned effect was directed towards protein degradation rather than mRNA stability. Plakophilin-2 expression decreases concurrently with the decline in CTNNA3 expression. Therefore, the expression of αT-catenin and plakophilin-2 could be potential surrogates for the diagnosis of ARVC.
Collapse
Affiliation(s)
- Pei-Fang Hung
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
| | - Fa-Po Chung
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Correspondence:
| | - Chung-Lieh Hung
- Department of Medicine, Mackay Medical College, New Taipei City 252005, Taiwan;
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252005, Taiwan
- Division of Cardiology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104217, Taiwan
| | - Yenn-Jiang Lin
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Tzu-Ting Kuo
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Jo-Nan Liao
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| | - Yun-Yu Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
- Institute of Epidemiology and Preventive Medicine College of Public Health, National Taiwan University, Taipei 100025, Taiwan
| | - Chih-Hsin Pan
- Institute of Forensic Medicine, Ministry of Justice, New Taipei City 235016, Taiwan; (C.-H.P.); (K.-P.S.)
| | - Kai-Ping Shaw
- Institute of Forensic Medicine, Ministry of Justice, New Taipei City 235016, Taiwan; (C.-H.P.); (K.-P.S.)
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan; (P.-F.H.); (Y.-J.L.); (J.-N.L.); (Y.-Y.C.); (S.-A.C.)
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
| |
Collapse
|
32
|
Arrhythmogenic Right Ventricular Cardiomyopathy. JACC Clin Electrophysiol 2022; 8:533-553. [PMID: 35450611 DOI: 10.1016/j.jacep.2021.12.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 01/21/2023]
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) encompasses a group of conditions characterized by right ventricular fibrofatty infiltration, with a predominant arrhythmic presentation. First described in the late 1970s and early 1980s, it is now frequently recognized to have biventricular involvement. The prevalence is ∼1:2,000 to 1:5,000, depending on geographic location, and it has a slight male predominance. The diagnosis of ARVC is determined on the basis of fulfillment of task force criteria incorporating electrophysiological parameters, cardiac imaging findings, genetic factors, and histopathologic features. Risk stratification of patients with ARVC aims to identify those who are at increased risk of sudden cardiac death or sustained ventricular tachycardia. Factors including age, sex, electrophysiological features, and cardiac imaging investigations all contribute to risk stratification. The current management of ARVC includes exercise restriction, β-blocker therapy, consideration for implantable cardioverter-defibrillator insertion, and catheter ablation. This review summarizes our current understanding of ARVC and provides clinicians with a practical approach to diagnosis and management.
Collapse
|
33
|
Lou J, Chen H, Huang S, Chen P, Yu Y, Chen F. Update on risk factors and biomarkers of sudden unexplained cardiac death. J Forensic Leg Med 2022; 87:102332. [DOI: 10.1016/j.jflm.2022.102332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 02/01/2023]
|
34
|
Chen SN, Lam CK, Wan YW, Gao S, Malak OA, Zhao SR, Lombardi R, Ambardekar AV, Bristow MR, Cleveland J, Gigli M, Sinagra G, Graw S, Taylor MR, Wu JC, Mestroni L. Activation of PDGFRA signaling contributes to filamin C-related arrhythmogenic cardiomyopathy. SCIENCE ADVANCES 2022; 8:eabk0052. [PMID: 35196083 PMCID: PMC8865769 DOI: 10.1126/sciadv.abk0052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/25/2021] [Indexed: 05/07/2023]
Abstract
FLNC truncating mutations (FLNCtv) are prevalent causes of inherited dilated cardiomyopathy (DCM), with a high risk of developing arrhythmogenic cardiomyopathy. We investigated the molecular mechanisms of mutant FLNC in the pathogenesis of arrhythmogenic DCM (a-DCM) using patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). We demonstrated that iPSC-CMs from two patients with different FLNCtv mutations displayed arrhythmias and impaired contraction. FLNC ablation induced a similar phenotype, suggesting that FLNCtv are loss-of-function mutations. Coimmunoprecipitation and proteomic analysis identified β-catenin (CTNNB1) as a downstream target. FLNC deficiency induced nuclear translocation of CTNNB1 and subsequently activated the platelet-derived growth factor receptor alpha (PDGFRA) pathway, which were also observed in human hearts with a-DCM and FLNCtv. Treatment with the PDGFRA inhibitor, crenolanib, improved contractile function of patient iPSC-CMs. Collectively, our findings suggest that PDGFRA signaling is implicated in the pathogenesis, and inhibition of this pathway is a potential therapeutic strategy in FLNC-related cardiomyopathies.
Collapse
Affiliation(s)
- Suet Nee Chen
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shanshan Gao
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Olfat A. Malak
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Raffaella Lombardi
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
- Department of Advanced Biomedical Sciences University of Naples “Federico II”, Naples, Italy
| | - Amrut V. Ambardekar
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Michael R. Bristow
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Joseph Cleveland
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Marta Gigli
- Cardiovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Sharon Graw
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Matthew R.G. Taylor
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Luisa Mestroni
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| |
Collapse
|
35
|
Pathology of sudden death, cardiac arrhythmias, and conduction system. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
36
|
Bueno-Beti C, Asimaki A. Histopathological Features and Protein Markers of Arrhythmogenic Cardiomyopathy. Front Cardiovasc Med 2021; 8:746321. [PMID: 34950711 PMCID: PMC8688541 DOI: 10.3389/fcvm.2021.746321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/17/2021] [Indexed: 11/13/2022] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a heritable heart muscle disease characterized by syncope, palpitations, ventricular arrhythmias and sudden cardiac death (SCD) especially in young individuals. It is estimated to affect 1:5,000 individuals in the general population, with >60% of patients bearing one or more mutations in genes coding for desmosomal proteins. Desmosomes are intercellular adhesion junctions, which in cardiac myocytes reside within the intercalated disks (IDs), the areas of mechanical and electrical cell-cell coupling. Histologically, ACM is characterized by fibrofatty replacement of cardiac myocytes predominantly in the right ventricular free wall though left ventricular and biventricular forms have also been described. The disease is characterized by age-related progression, vast phenotypic manifestation and incomplete penetrance, making proband diagnosis and risk stratification of family members particularly challenging. Key protein redistribution at the IDs may represent a specific diagnostic marker but its applicability is still limited by the need for a myocardial sample. Specific markers of ACM in surrogate tissues, such as the blood and the buccal epithelium, may represent a non-invasive, safe and inexpensive alternative for diagnosis and cascade screening. In this review, we shall cover the most relevant biomarkers so far reported and discuss their potential impact on the diagnosis, prognosis and management of ACM.
Collapse
Affiliation(s)
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St. George's University of London, London, United Kingdom
| |
Collapse
|
37
|
Przybylski R, Abrams DJ. Clinical and genetic features of arrhythmogenic cardiomyopathy: the electrophysiology perspective. PROGRESS IN PEDIATRIC CARDIOLOGY 2021. [DOI: 10.1016/j.ppedcard.2021.101463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
38
|
MicroRNAs: From Junk RNA to Life Regulators and Their Role in Cardiovascular Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) are single-stranded small non-coding RNA (18–25 nucleotides) that until a few years ago were considered junk RNA. In the last twenty years, they have acquired more importance thanks to the understanding of their influence on gene expression and their role as negative regulators at post-transcriptional level, influencing the stability of messenger RNA (mRNA). Approximately 5% of the genome encodes miRNAs which are responsible for regulating numerous signaling pathways, cellular processes and cell-to-cell communication. In the cardiovascular system, miRNAs control the functions of various cells, such as cardiomyocytes, endothelial cells, smooth muscle cells and fibroblasts, playing a role in physiological and pathological processes and seeming also related to variations in contractility and hereditary cardiomyopathies. They provide a new perspective on the pathophysiology of disorders such as hypertrophy, fibrosis, arrhythmia, inflammation and atherosclerosis. MiRNAs are differentially expressed in diseased tissue and can be released into the circulation and then detected. MiRNAs have become interesting for the development of new diagnostic and therapeutic tools for various diseases, including heart disease. In this review, the concept of miRNAs and their role in cardiomyopathies will be introduced, focusing on their potential as therapeutic and diagnostic targets (as biomarkers).
Collapse
|
39
|
Moazzen H, Venger K, Kant S, Leube RE, Krusche CA. Desmoglein 2 regulates cardiogenesis by restricting hematopoiesis in the developing murine heart. Sci Rep 2021; 11:21687. [PMID: 34737300 PMCID: PMC8569146 DOI: 10.1038/s41598-021-00996-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/21/2021] [Indexed: 02/05/2023] Open
Abstract
Cardiac morphogenesis relies on intricate intercellular signaling. Altered signaling impacts cardiac function and is detrimental to embryonic survival. Here we report an unexpected regulatory role of the desmosomal cell adhesion molecule desmoglein 2 (Dsg2) on murine heart development. A large percentage of Dsg2-mutant embryos develop pericardial hemorrhage. Lethal myocardial rupture is occasionally observed, which is not associated with loss of cardiomyocyte contact but with expansion of abnormal, non-myocyte cell clusters within the myocardial wall. Two types of abnormal cell clusters can be distinguished: Type A clusters involve endocard-associated, round-shaped CD31+ cells, which proliferate and invade the myocardium. They acquire Runx1- and CD44-positivity indicating a shift towards a hematopoietic phenotype. Type B clusters expand subepicardially and next to type A clusters. They consist primarily of Ter119+ erythroid cells with interspersed Runx1+/CD44+ cells suggesting that they originate from type A cell clusters. The observed pericardial hemorrhage is caused by migration of erythrocytes from type B clusters through the epicardium and rupture of the altered cardiac wall. Finally, evidence is presented that structural defects of Dsg2-depleted cardiomyocytes are primary to the observed pathogenesis. We propose that cardiomyocyte-driven paracrine signaling, which likely involves Notch1, directs subsequent trans-differentiation of endo- and epicardial cells. Together, our observations uncover a hitherto unknown regulatory role of Dsg2 in cardiogenesis.
Collapse
Affiliation(s)
- Hoda Moazzen
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Kateryna Venger
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
40
|
Ng KE, Delaney PJ, Thenet D, Murtough S, Webb CM, Zaman N, Tsisanova E, Mastroianni G, Walker SLM, Westaby JD, Pennington DJ, Pink R, Kelsell DP, Tinker A. Early inflammation precedes cardiac fibrosis and heart failure in desmoglein 2 murine model of arrhythmogenic cardiomyopathy. Cell Tissue Res 2021; 386:79-98. [PMID: 34236518 PMCID: PMC8526453 DOI: 10.1007/s00441-021-03488-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 06/18/2021] [Indexed: 12/19/2022]
Abstract
The study of a desmoglein 2 murine model of arrhythmogenic cardiomyopathy revealed cardiac inflammation as a key early event leading to fibrosis. Arrhythmogenic cardiomyopathy (AC) is an inherited heart muscle disorder leading to ventricular arrhythmias and heart failure due to abnormalities in the cardiac desmosome. We examined how loss of desmoglein 2 (Dsg2) in the young murine heart leads to development of AC. Apoptosis was an early cellular phenotype, and RNA sequencing analysis revealed early activation of inflammatory-associated pathways in Dsg2-null (Dsg2-/-) hearts at postnatal day 14 (2 weeks) that were absent in the fibrotic heart of adult mice (10 weeks). This included upregulation of iRhom2/ADAM17 and its associated pro-inflammatory cytokines and receptors such as TNFα, IL6R and IL-6. Furthermore, genes linked to specific macrophage populations were also upregulated. This suggests cardiomyocyte stress triggers an early immune response to clear apoptotic cells allowing tissue remodelling later on in the fibrotic heart. Our analysis at the early disease stage suggests cardiac inflammation is an important response and may be one of the mechanisms responsible for AC disease progression.
Collapse
Affiliation(s)
- K E Ng
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.,Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - P J Delaney
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - D Thenet
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - S Murtough
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - C M Webb
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - N Zaman
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - E Tsisanova
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - G Mastroianni
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - S L M Walker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - J D Westaby
- CRY Dept. of Cardiovascular Pathology, Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Institute, St. George's University of London, Jenner WingCranmer Terrace, London, SW17 0RE, UK
| | - D J Pennington
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - R Pink
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Headington Campus, Oxford, OX3 0BP, UK
| | - D P Kelsell
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - A Tinker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
41
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Shin Kadota
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yan Shen
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences Shinshu University Matsumoto, Nagano 390-8621 Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR) Kyoto University Uji Kyoto 611-0011 Japan
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS) Kyoto University Kyoto 606-8501 Japan
- School of Pharmacy Fudan University Shanghai 201203 China
| |
Collapse
|
42
|
Cheedipudi SM, Fan S, Rouhi L, Marian AJ. Pharmacological suppression of the WNT signaling pathway attenuates age-dependent expression of the phenotype in a mouse model of arrhythmogenic cardiomyopathy. THE JOURNAL OF CARDIOVASCULAR AGING 2021; 1. [PMID: 34447973 PMCID: PMC8386676 DOI: 10.20517/jca.2021.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction Arrhythmogenic cardiomyopathy (ACM) is a genetic disease of the myocardium, characterized by cardiac arrhythmias, dysfunction, and sudden cardiac death. The pathological hallmark of ACM is fibro-adipocytes replacing cardiac myocytes. The canonical WNT pathway is implicated in the pathogenesis of ACM. Aim The study aimed to determine the effects of the suppression of the WNT pathway on cardiac phenotype in a mouse model of ACM. Methods and Results One copy of the Dsp gene, a known cause of ACM in humans, was deleted specifically in cardiac myocytes (Myh6-Cre-Dsp W/F). Three-month-old wild type and Myh6-Cre-Dsp W/F mice, without a discernible phenotype, were randomized to either untreated or daily administration of a vehicle (placebo), or WNT974, the latter an established inhibitor of the WNT pathway, for three months. The Myh6-Cre-Dsp W/F mice in the untreated or placebo-treated groups exhibited cardiac dilatation and dysfunction, increased myocardial fibrosis, and apoptosis upon completion of the study, which was verified by complementary methods. Daily administration of WNT974 prevented and/or attenuated evolving cardiac dilatation and dysfunction, normalized myocardial fibrosis, and reduced apoptosis, compared to the untreated or placebo-treated groups. However, administration of WNT974 increased the number of adipocytes only in the Myh6-Cre-Dsp W/F hearts. There were no differences in the incidence of cardiac arrhythmias and survival rates. Conclusion Suppression of the WNT pathway imparts salutary phenotypic effects by preventing or attenuating age-dependent expression of cardiac dilatation and dysfunction, myocardial fibrosis, and apoptosis in a mouse model of ACM. The findings set the stage for large-scale studies and studies in larger animal models to test the beneficial effects of the suppression of the WNT pathway in ACM. One sentence summary Suppression of the WNT signaling pathway has beneficial effects on cardiac dysfunction, myocardial apoptosis, and fibrosis in a mouse model of arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Sirisha M Cheedipudi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Siyang Fan
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Leila Rouhi
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| | - Ali J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
43
|
Simons E, Labro A, Saenen J, Nijak A, Sieliwonczyk E, Vandendriessche B, Dąbrowska M, Van Craenenbroeck EM, Schepers D, Van Laer L, Loeys BL, Alaerts M. Molecular autopsy and subsequent functional analysis reveal de novo DSG2 mutation as cause of sudden death. Eur J Med Genet 2021; 64:104322. [PMID: 34438094 DOI: 10.1016/j.ejmg.2021.104322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/15/2021] [Accepted: 08/22/2021] [Indexed: 11/28/2022]
Abstract
Sudden cardiac death (SCD) is a common cause of death in young adults. In up to 80% of cases a genetic cause is suspected. Next-generation sequencing of candidate genes can reveal the cause of SCD, provide prognostic management, and facilitate pre-symptomatic testing and prevention in relatives. Here we present a proband who experienced SCD in his sleep for which molecular autopsy was performed. We performed a post-mortem genetic analysis of a 49-year-old male who died during sleep after competitive kayaking, using a Cardiomyopathy and Primary Arrhythmia next-generation sequencing panel, each containing 51 candidate genes. Autopsy was not performed. Genetic testing of the proband resulted in missense variants in KCNQ1 (c.1449C > A; p.(Asn483Lys)) and DSG2 (c.2979G > T; p.(Gln993His)), both absent from the gnomAD database. Familial segregation analysis showed de novo occurrence of the DSG2 variant and presence of the KCNQ1 variant in the proband's mother and daughter. KCNQ1 p.(Asn483Lys) was predicted to be pathogenic by MutationTaster. However, none of the KCNQ1 variant carrying family members showed long QTc on ECG or Holter. We further functionally analysed this variant using patch-clamp in a heterologous expression system (Chinese Hamster Ovary (CHO) cells) expressing the KCNQ1 mutant in combination with KCNE1 wild type protein and showed no significant changes in electrophysiological function of Kv7.1. Based on the above evidence, we concluded that the DSG2 p.(Gln993His) variant is the most likely cause of SCD in the presented case, and that there is insufficient evidence that the identified KCNQ1 p.(Asn483Lys) variant would confer risk for SCD in his mother and daughter. Fortunately, the DSG2 variant was not inherited by the proband's two children. This case report indicates the added value of molecular autopsy and the importance of subsequent functional study of variants to inform patients and family members about the risk of variants they might carry.
Collapse
Affiliation(s)
- Eline Simons
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Alain Labro
- Laboratory for Molecular, Cellular and Network Excitability, University of Antwerp, Antwerp, Belgium; Department of Basic and Applied Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Johan Saenen
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Aleksandra Nijak
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Ewa Sieliwonczyk
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bert Vandendriessche
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Małgorzata Dąbrowska
- Laboratory for Molecular, Cellular and Network Excitability, University of Antwerp, Antwerp, Belgium
| | | | - Dorien Schepers
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Lut Van Laer
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Bart L Loeys
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Maaike Alaerts
- Cardiogenetics Research Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|
44
|
Takemoto Y, Kadota S, Minami I, Otsuka S, Okuda S, Abo M, Punzalan LL, Shen Y, Shiba Y, Uesugi M. Chemical Genetics Reveals a Role of Squalene Synthase in TGFβ Signaling and Cardiomyogenesis. Angew Chem Int Ed Engl 2021; 60:21824-21831. [PMID: 34374184 DOI: 10.1002/anie.202100523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Indexed: 11/11/2022]
Abstract
KY02111 is a widely used small molecule that boosts cardiomyogenesis of the mesoderm cells derived from pluripotent stem cells, yet its molecular mechanism of action remains elusive. The present study resolves the initially perplexing effects of KY02111 on Wnt signaling and subsequently identifies squalene synthase (SQS) as a molecular target of KY02111 and its optimized version, KY-I. By disrupting the interaction of SQS with cardiac ER-membrane protein TMEM43, KY02111 impairs TGFβ signaling, but not Wnt signaling, and thereby recapitulates the clinical mutation of TMEM43 that causes arrhythmogenic right ventricular cardiomyopathy (ARVC), an inherited heart disease that involves a substitution of myocardium with fatty tissue. These findings reveal a heretofore undescribed role of SQS in TGFβ signaling and cardiomyogenesis. KY02111 may find its use in ARVC modeling as well as serve as a chemical tool for studying TGFβ/SMAD signaling.
Collapse
Affiliation(s)
- Yasushi Takemoto
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Shin Kadota
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Itsunari Minami
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan
| | - Shinya Otsuka
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Satoshi Okuda
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Masahiro Abo
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Louvy Lynn Punzalan
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yan Shen
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Yuji Shiba
- Institute for Biomedical Sciences, Shinshu University, Matsumoto, Nagano, 390-8621, Japan
| | - Motonari Uesugi
- Institute for Chemical Research (ICR), Kyoto University, Uji, Kyoto 611-0011, Japan.,Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto, 606-8501, Japan.,School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
45
|
Kukavica D, Trancuccio A, Arnò C, Latini AC, Mazzanti A, Priori SG. Desmoplakin cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy: two distinct forms of cardiomyopathy? Minerva Cardiol Angiol 2021; 70:217-237. [PMID: 34338490 DOI: 10.23736/s2724-5683.21.05804-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The confirmation of a hypothesis that desmoplakin-related (DSP) cardiomyopathy could represent a distinct clinical entity from the classical, RV-dominant, form of arrhythmogenic cardiomyopathy (ACM), most frequently caused by PKP2 mutations, would without any shadow of doubt signify a turning point in the history of this disease. The concept of gene-specific diseases underneath the umbrella diagnosis of ACM would bring fundamental changes not only in the clinical, diagnostic and therapeutic approach, but also in terms of risk stratification, pushing the scientific community towards a more patient-centred view of the disease, similarly to what has already been done in other inherited arrhythmogenic disease (e.g., Long QT Syndrome; LQTS). We provide a state-of-the-art review, starting with a brief historical framework to give the necessary context and better focus the question. Then, we proceed with a novel, genotype-tophenotype-based comparison of the most important aspects of DSP-related cardiomyopathy with the classical, RV-dominant ACM: this allows us to ascertain not only that the differences between the forms exist, but are also clinically relevant and actionable, leading to the underrecognition of the atypical, DSP-related, LV-dominant forms when applying the current diagnostic criteria. These findings will usher an exciting era, in which the scientific community will try to answer a range of questions, starting from the reasons why different desmosomal mutations cause such different phenotypes.
Collapse
Affiliation(s)
- Deni Kukavica
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Molecular Cardiology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Alessandro Trancuccio
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Molecular Cardiology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Carlo Arnò
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
| | - Alessia C Latini
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy
| | - Andrea Mazzanti
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Molecular Cardiology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Silvia G Priori
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy - .,Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Molecular Cardiology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
46
|
Chelko SP, Keceli G, Carpi A, Doti N, Agrimi J, Asimaki A, Beti CB, Miyamoto M, Amat-Codina N, Bedja D, Wei AC, Murray B, Tichnell C, Kwon C, Calkins H, James CA, O'Rourke B, Halushka MK, Melloni E, Saffitz JE, Judge DP, Ruvo M, Kitsis RN, Andersen P, Di Lisa F, Paolocci N. Exercise triggers CAPN1-mediated AIF truncation, inducing myocyte cell death in arrhythmogenic cardiomyopathy. Sci Transl Med 2021; 13:13/581/eabf0891. [PMID: 33597260 DOI: 10.1126/scitranslmed.abf0891] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
Myocyte death occurs in many inherited and acquired cardiomyopathies, including arrhythmogenic cardiomyopathy (ACM), a genetic heart disease plagued by the prevalence of sudden cardiac death. Individuals with ACM and harboring pathogenic desmosomal variants, such as desmoglein-2 (DSG2), often show myocyte necrosis with progression to exercise-associated heart failure. Here, we showed that homozygous Dsg2 mutant mice (Dsg2 mut/mut), a model of ACM, die prematurely during swimming and display myocardial dysfunction and necrosis. We detected calcium (Ca2+) overload in Dsg2 mut/mut hearts, which induced calpain-1 (CAPN1) activation, association of CAPN1 with mitochondria, and CAPN1-induced cleavage of mitochondrial-bound apoptosis-inducing factor (AIF). Cleaved AIF translocated to the myocyte nucleus triggering large-scale DNA fragmentation and cell death, an effect potentiated by mitochondrial-driven AIF oxidation. Posttranslational oxidation of AIF cysteine residues was due, in part, to a depleted mitochondrial thioredoxin-2 redox system. Hearts from exercised Dsg2 mut/mut mice were depleted of calpastatin (CAST), an endogenous CAPN1 inhibitor, and overexpressing CAST in myocytes protected against Ca2+ overload-induced necrosis. When cardiomyocytes differentiated from Dsg2 mut/mut embryonic stem cells (ES-CMs) were challenged with β-adrenergic stimulation, CAPN1 inhibition attenuated CAPN1-induced AIF truncation. In addition, pretreatment of Dsg2 mut/mut ES-CMs with an AIF-mimetic peptide, mirroring the cyclophilin-A (PPIA) binding site of AIF, blocked PPIA-mediated AIF-nuclear translocation, and reduced both apoptosis and necrosis. Thus, preventing CAPN1-induced AIF-truncation or barring binding of AIF to the nuclear chaperone, PPIA, may avert myocyte death and, ultimately, disease progression to heart failure in ACM and likely other forms of cardiomyopathies.
Collapse
Affiliation(s)
- Stephen P Chelko
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA. .,Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrea Carpi
- Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, CNR, Naples 80134, Italy
| | - Jacopo Agrimi
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London WC1E 6BS, UK
| | - Carlos Bueno Beti
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London WC1E 6BS, UK
| | - Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Nuria Amat-Codina
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Australian School of Advanced Medicine, Macquarie University, Sydney, NSW 2109, Australia
| | - An-Chi Wei
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Crystal Tichnell
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hugh Calkins
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Cynthia A James
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Edon Melloni
- Department of Medicine, University of Genova, Genova 16126, Italy
| | - Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 20115, USA
| | - Daniel P Judge
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Medical University of South Carolina, Charleston, SC 29425, USA
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, CNR, Naples 80134, Italy
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA. .,Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| |
Collapse
|
47
|
Migliore F, Mattesi G, Zorzi A, Bauce B, Rigato I, Corrado D, Cipriani A. Arrhythmogenic Cardiomyopathy-Current Treatment and Future Options. J Clin Med 2021; 10:2750. [PMID: 34206637 PMCID: PMC8268983 DOI: 10.3390/jcm10132750] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inheritable heart muscle disease characterised pathologically by fibrofatty myocardial replacement and clinically by ventricular arrhythmias (VAs) and sudden cardiac death (SCD). Although, in its original description, the disease was believed to predominantly involve the right ventricle, biventricular and left-dominant variants, in which the myocardial lesions affect in parallel or even mostly the left ventricle, are nowadays commonly observed. The clinical management of these patients has two main purposes: the prevention of SCD and the control of arrhythmic and heart failure (HF) events. An implantable cardioverter defibrillator (ICD) is the only proven lifesaving treatment, despite significant morbidity because of device-related complications and inappropriate shocks. Selection of patients who can benefit the most from ICD therapy is one of the most challenging issues in clinical practice. Risk stratification in ACM patients is mostly based on arrhythmic burden and ventricular dysfunction severity, although other clinical features resulting from electrocardiogram and imaging modalities such as cardiac magnetic resonance may have a role. Medical therapy is crucial for treatment of VAs and the prevention of negative ventricular remodelling. In this regard, the efficacy of novel anti-HF molecules and drugs acting on the inflammatory pathway in patients with ACM is, to date, unknown. Catheter ablation represents an effective strategy to treat ventricular tachycardia relapses and recurrent ICD shocks. The present review will address the current strategies for prevention of SCD and treatment of VAs and HF in patients with ACM.
Collapse
Affiliation(s)
- Federico Migliore
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Giulia Mattesi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alessandro Zorzi
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Barbara Bauce
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Ilaria Rigato
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Domenico Corrado
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Alberto Cipriani
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| |
Collapse
|
48
|
Lao N, Laiq Z, Courson J, Al-Quthami A. Left-dominant arrhythmogenic cardiomyopathy: an association with desmoglein-2 gene mutation-a case report. Eur Heart J Case Rep 2021; 5:ytab213. [PMID: 34263121 PMCID: PMC8274644 DOI: 10.1093/ehjcr/ytab213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/13/2021] [Accepted: 05/06/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Desmosomes are specialized intercellular adhesive junctions of cardiac and epithelial cells that provide intercellular mechanical coupling through glycoproteins, one of which is desmoglein (DSG). DSG-2 mutations are frequently associated with biventricular arrhythmogenic cardiomyopathy (ACM). We report a case of left-dominant ACM in a patient who initially was misclassified as dilated cardiomyopathy (DCM). CASE SUMMARY A 28-year-old-woman was found to have a moderately reduced left ventricular (LV) systolic function and frequent premature ventricular contractions (PVCs). Targeted genetic testing revealed a heterozygous likely pathogenic variant associated with ACM in exon 15 of the DSG-2 gene (c.3059_3062del; p.Glu1020Alafs*18). Subsequent cardiac magnetic resonance (CMR) imaging showed epicardial and mid-myocardial fatty infiltration involving multiple LV wall segments, multiple areas of mid-myocardial fibrosis/scar, regional dyskinesis involving both ventricles, and an overall reduced left ventricular ejection fraction. The patient's right ventricular (RV) cavity size and overall RV systolic function were normal. Based on the patient's frequent PVCs, family history, fibrofatty myocardial replacement in multiple LV segments, and dyskinetic motion of multiple ventricular wall segments (predominantly affecting the LV), the patient was diagnosed with left-dominant ACM. DISCUSSION Identifying a likely pathogenic mutation associated with ACM in a patient with ventricular arrhythmias and a family history of sudden cardiac death increased the possibility of ACM. Subsequent CMR imaging confirmed the diagnosis of left-dominant ACM by demonstrating regional biventricular dyskinesia and a characteristic pattern of fibrofatty myocardial replacement. Our case highlights the importance of targeted genetic testing and advanced cardiac imaging in distinguishing left-dominant ACM from DCM.
Collapse
Affiliation(s)
- Nicole Lao
- Department of Internal Medicine, Cleveland Clinic Akron General, Akron, 1 Akron General Ave, Akron, OH, USA 44307, USA
| | - Zenab Laiq
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Akron General, Akron, 1 Akron General Ave, Akron, OH, USA 44307, USA
| | - Jeffrey Courson
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Akron General, Akron, 1 Akron General Ave, Akron, OH, USA 44307, USA
| | - Adeeb Al-Quthami
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic Akron General, Akron, 1 Akron General Ave, Akron, OH, USA 44307, USA
| |
Collapse
|
49
|
Variants in MHY7 Gene Cause Arrhythmogenic Cardiomyopathy. Genes (Basel) 2021; 12:genes12060793. [PMID: 34067482 PMCID: PMC8224781 DOI: 10.3390/genes12060793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Arrhythmogenic Cardiomyopathy (ACM) is a disease of the cardiac muscle, characterized by frequent ventricular arrhythmias and functional/ structural abnormalities, mainly of the right ventricle. To date, 20 different genes have been associated with ACM and the majority of them encode for desmosomal proteins. In this study, we describe the characterization of two novel variants in MHY7 gene, segregating in two ACM families. MYH7 encodes for myosin heavy chain β (MHC-β) isoform, involved in cardiac muscle contractility. METHOD AND RESULTS In family A, the autopsy revealed ACM with biventricular involvement in both the proband and his father. In family B, the proband had been diagnosed as affected by ACM and implanted with implantable cardioverter defibrillator (ICD), due to ECG evidence of monomorphic ventricular tachycardia after syncope. After clinical evaluation, a molecular diagnosis was performed using a NGS custom panel. The two novel variants identified predicted damaging, located in a highly conserved domain: c. 2630T>C is not described while c.2609G>A has a frequency of 0.00000398. In silico analyses evaluated the docking characteristics between proteins using the Haddock2.2 webserver. CONCLUSIONS Our results reveal two variants in sarcomeric genes to be the molecular cause of ACM, further increasing the genetic heterogeneity of the disease; in fact, sarcomeric variants are usually associated with HCM phenotype. Studies on the role of sarcomere genes in the pathogenesis of ACM are surely recommended in those ACM patients negative for desmosomal mutation screening.
Collapse
|
50
|
Huang R, Luo Y, Zhao J, Su K, Lei Y, Li Y. Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC) Probably Caused by DSG2 p.Val149Ile Mutation as Genetic Background When Carrying with Heterozygous PRRT2 p.Arg217ProfsTer8 Mutation: A Case Report. Int Med Case Rep J 2021; 14:307-313. [PMID: 34012299 PMCID: PMC8128131 DOI: 10.2147/imcrj.s309668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background ARVC is a rare genetic-related disease characterized by fibrous fat replacement in the ventricular myocardium, caused by mutations in genes encoding for the desmosomal proteins, such as the desmoglein-2 gene (DSG2). It is reported in the literature that other genetic factors may play a role in disease penetrance. Herein, we report a Chinese proband with ARVC, which was probably caused by DSG2 p.Val149Ile mutation as genetic background when carrying heterozygous PRRT2 p.Arg217ProfsTer8 mutation. Case Presentation A 17-year-old male with a history of paroxysmal kinesigenic dyskinesia (PKD) presented to the hospital for syncope induced by ventricular tachycardia. According to relevant clinical data and the diagnostic criteria of ARVC, a precise positive diagnosis of ARVC was finally made. Gene testing revealed that the patient carried a DSG2 heterozygous missense mutation (NM_001943: exon5: c.445G>A, p.Val149Ile) as well as frameshift mutation of PRRT2 (NM_001256442: exon2: p. Arg217Profs Ter8). Conclusion This is the first time to report a Chinese proband with ARVC and a history of PKD carrying both DSG2 p. val149ile mutation and PRRT2 p. Arg217ProfsTer8 mutation, which can provide a new direction for gene screening of patients with ARVC and further supplements for its diagnostic criteria.
Collapse
Affiliation(s)
- Rui Huang
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi Prefecture, 445000, Hubei Province, People's Republic of China
| | - YinHua Luo
- Department of Central Hospital of Tujia and Miao Autonomous Prefecture, Hubei University of Medicine, Shiyan, 442000, Hubei Province, People's Republic of China
| | - Jingbo Zhao
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi Prefecture, 445000, Hubei Province, People's Republic of China
| | - Ke Su
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi Prefecture, 445000, Hubei Province, People's Republic of China
| | - YuHua Lei
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi Prefecture, 445000, Hubei Province, People's Republic of China
| | - Yuanhong Li
- Cardiovascular Disease Center, Central Hospital of Tujia and Miao Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi Prefecture, 445000, Hubei Province, People's Republic of China
| |
Collapse
|